1
|
Mangarova DB, Reimann C, Kaufmann JO, Möckel J, Kader A, Adams LC, Ludwig A, Onthank D, Robinson S, Karst U, Helmer R, Botnar R, Hamm B, Makowski MR, Brangsch J. Elastin-specific MR probe for visualization and evaluation of an interleukin-1β targeted therapy for atherosclerosis. Sci Rep 2024; 14:20648. [PMID: 39232217 PMCID: PMC11375012 DOI: 10.1038/s41598-024-71716-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory condition of the arteries and represents the primary cause of various cardiovascular diseases. Despite ongoing progress, finding effective anti-inflammatory therapeutic strategies for atherosclerosis remains a challenge. Here, we assessed the potential of molecular magnetic resonance imaging (MRI) to visualize the effects of 01BSUR, an anti-interleukin-1β monoclonal antibody, for treating atherosclerosis in a murine model. Male apolipoprotein E-deficient mice were divided into a therapy group (01BSUR, 2 × 0.3 mg/kg subcutaneously, n = 10) and control group (no treatment, n = 10) and received a high-fat diet for eight weeks. The plaque burden was assessed using an elastin-targeted gadolinium-based contrast probe (0.2 mmol/kg intravenously) on a 3 T MRI scanner. T1-weighted imaging showed a significantly lower contrast-to-noise (CNR) ratio in the 01BSUR group (pre: 3.93042664; post: 8.4007067) compared to the control group (pre: 3.70679168; post: 13.2982156) following administration of the elastin-specific MRI probe (p < 0.05). Histological examinations demonstrated a significant reduction in plaque size (p < 0.05) and a significant decrease in plaque elastin content (p < 0.05) in the treatment group compared to control animals. This study demonstrated that 01BSUR hinders the progression of atherosclerosis in a mouse model. Using an elastin-targeted MRI probe, we could quantify these therapeutic effects in MRI.
Collapse
Affiliation(s)
- Dilyana Branimirova Mangarova
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Carolin Reimann
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Jan Ole Kaufmann
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing (BAM), Richard-Willstätter-Str. 11, 12489, Berlin, Germany
| | - Jana Möckel
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Avan Kader
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Lisa Christine Adams
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Antje Ludwig
- Department of Cardiology and Angiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Berlin, Germany
| | - David Onthank
- Lantheus Medical Imaging, 331 Treble Cove Road, North Billerica, MA, United States of America
| | - Simon Robinson
- Lantheus Medical Imaging, 331 Treble Cove Road, North Billerica, MA, United States of America
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Rebecca Helmer
- Institute of Inorganic and Analytical Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Rene Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital Westminster Bridge Road, London, SE1 7EH, United Kingdom
- Wellcome Trust/EPSRC Centre for Medical Engineering, King's College London, London, United Kingdom
- BHF Centre of Excellence, King's College London, Denmark Hill Campus, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
- Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bernd Hamm
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus Richard Makowski
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Julia Brangsch
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
2
|
Zhou S, Ma B, Luo M. Matrix metalloproteinases in aortic dissection. Vascul Pharmacol 2024; 156:107420. [PMID: 39182633 DOI: 10.1016/j.vph.2024.107420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Aortic dissection, characterized by a high immediate mortality, is primarily caused by excessive bleeding within the walls of the aorta or a severe tear within the intimal layer of the aorta. Inflammation, as well as oxidative stress and the degradation of extracellular matrix (ECM), are significant factors in the development and occurrence of aortic dissection. Matrix metalloproteinases (MMPs) are pivotal enzymes responsible for degrading the ECM. Inflammatory factors and oxidants can interact with MMPs, indicating the potential significance of MMPs in aortic dissection. A substantial body of evidence indicates that numerous MMPs are significantly upregulated in aortic dissection, playing a critical role in ECM degradation and the pathogenesis of aortic dissection. Furthermore, targeting these enzymes has demonstrated potential in facilitating ECM restoration and reducing the incidence of aortic dissection. This review initially provides a brief overview of MMP biology before delving into their expression patterns, regulatory mechanisms, and therapeutic applications in aortic dissection. A profound comprehension of the catabolic pathways associated with aortic dissection is imperative for the future development of potential preventive or therapeutic bio-interventions for aortic dissection.
Collapse
Affiliation(s)
- Shufen Zhou
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Baihui Ma
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Mingyao Luo
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Department of Vascular Surgery, Central-China Branch of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China; Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650102, China.
| |
Collapse
|
3
|
Wu Z, Zhang P, Yue J, Wang Q, Zhuang P, Jehan S, Fan L, Xue J, Zhou W, Wang H. Tea polyphenol nanoparticles enable targeted siRNA delivery and multi-bioactive therapy for abdominal aortic aneurysms. J Nanobiotechnology 2024; 22:471. [PMID: 39118143 PMCID: PMC11308685 DOI: 10.1186/s12951-024-02756-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease, while there is a lack of pharmaceutical interventions to halt AAA progression presently. To address the multifaceted pathology of AAA, this work develops a novel multifunctional gene delivery system to simultaneously deliver two siRNAs targeting MMP-2 and MMP-9. The system (TPNs-siRNA), formed through the oxidative polymerization and self-assembly of epigallocatechin gallate (EGCG), efficiently encapsulates siRNAs during self-assembly. TPNs-siRNA safeguards siRNAs from biological degradation, facilitates intracellular siRNA transfection, promotes lysosomal escape, and releases siRNAs to silence MMP-2 and MMP-9. Additionally, TPNs, serving as a multi-bioactive material, mitigates oxidative stress and inflammation, fosters M1-to-M2 repolarization of macrophages, and inhibits cell calcification and apoptosis. In experiments with AAA mice, TPNs-siRNA accumulated and persisted in aneurysmal tissue after intravenous delivery, demonstrating that TPNs-siRNA can be significantly distributed in macrophages and VSMCs relevant to AAA pathogenesis. Leveraging the carrier's intrinsic multi-bioactive properties, the targeted siRNA delivery by TPNs exhibits a synergistic effect for enhanced AAA therapy. Furthermore, TPNs-siRNA is gradually metabolized and excreted from the body, resulting in excellent biocompatibility. Consequently, TPNs emerges as a promising multi-bioactive nanotherapy and a targeted delivery nanocarrier for effective AAA therapy.
Collapse
Affiliation(s)
- Zhen Wu
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Peng Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jie Yue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Qingshan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Peipei Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Shah Jehan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Liyuan Fan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jiarun Xue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Haiyang Wang
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
4
|
The mechanism and therapy of aortic aneurysms. Signal Transduct Target Ther 2023; 8:55. [PMID: 36737432 PMCID: PMC9898314 DOI: 10.1038/s41392-023-01325-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Aortic aneurysm is a chronic aortic disease affected by many factors. Although it is generally asymptomatic, it poses a significant threat to human life due to a high risk of rupture. Because of its strong concealment, it is difficult to diagnose the disease in the early stage. At present, there are no effective drugs for the treatment of aneurysms. Surgical intervention and endovascular treatment are the only therapies. Although current studies have discovered that inflammatory responses as well as the production and activation of various proteases promote aortic aneurysm, the specific mechanisms remain unclear. Researchers are further exploring the pathogenesis of aneurysms to find new targets for diagnosis and treatment. To better understand aortic aneurysm, this review elaborates on the discovery history of aortic aneurysm, main classification and clinical manifestations, related molecular mechanisms, clinical cohort studies and animal models, with the ultimate goal of providing insights into the treatment of this devastating disease. The underlying problem with aneurysm disease is weakening of the aortic wall, leading to progressive dilation. If not treated in time, the aortic aneurysm eventually ruptures. An aortic aneurysm is a local enlargement of an artery caused by a weakening of the aortic wall. The disease is usually asymptomatic but leads to high mortality due to the risk of artery rupture.
Collapse
|
5
|
Zhao Y, Hong X, Xie X, Guo D, Chen B, Fu W, Wang L. Preoperative systemic inflammatory response index predicts long-term outcomes in type B aortic dissection after endovascular repair. Front Immunol 2022; 13:992463. [PMID: 36248781 PMCID: PMC9554789 DOI: 10.3389/fimmu.2022.992463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Objectives Inflammation is a hallmark of the initial development and progression of aortic dissection. This study aimed to investigate the value of preoperative inflammatory biomarkers in predicting aorta-related adverse events (AAEs) after thoracic endovascular aortic repair (TEVAR) for type B aortic dissection. Methods We included all patients who underwent TEVAR for type B aortic dissection between November 2016 and November 2020 in this single-center, retrospective cohort study. Patients were divided into two groups: the AAEs group (n = 75) and the non-AAEs group (n = 126). Preoperative inflammatory biomarkers were recorded, including neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), systemic immune inflammation index (SII), and systemic inflammatory response index (SIRI). Patients were followed-up for the development of AAEs. Prediction accuracy of inflammatory biomarkers for AAEs were evaluated using the area under the receiver operating characteristic curves. Results This study included 201 patients, of whom 80.0% were men, with a mean age of 59.1 ± 12.5 years. A total of 75 patients developed AAEs after TEVAR. The AUCs of NLR, MLR, PLR, SII, and SIRI for AAEs were.746,.782,.534,.625 and.807, respectively. Age and SIRI were independent risk factors for the AAEs after TEVAR (HR 3.264, p <.001; HR 4.281, p <.001, respectively). Survival analysis revealed significantly lower AAE-free status in patients with preoperative SIRI > = 4 (p <.001). Conclusion Increased preoperative SIRI and age are independent risk factors for AAEs after TEVAR in type B aortic dissection.
Collapse
Affiliation(s)
- Yufei Zhao
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Xiang Hong
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Vascular Surgery, Fudan University, Shanghai, China
| | - Xinsheng Xie
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Vascular Surgery, Fudan University, Shanghai, China
| | - Daqiao Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Bin Chen
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Institute of Vascular Surgery, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- *Correspondence: Lixin Wang, ; Weiguo Fu,
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Institute of Vascular Surgery, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- *Correspondence: Lixin Wang, ; Weiguo Fu,
| |
Collapse
|
6
|
Puchenkova OA, Soldatov VO, Belykh AE, Bushueva O, Piavchenko GA, Venediktov AA, Shakhpazyan NK, Deykin AV, Korokin MV, Pokrovskiy MV. Cytokines in Abdominal Aortic Aneurysm: Master Regulators With Clinical Application. Biomark Insights 2022; 17:11772719221095676. [PMID: 35492378 PMCID: PMC9052234 DOI: 10.1177/11772719221095676] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 04/04/2022] [Indexed: 01/05/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a potentially life-threatening disorder with a mostly asymptomatic course where the abdominal aorta is weakened and bulged. Cytokines play especially important roles (both positive and negative) among the molecular actors of AAA development. All the inflammatory cascades, extracellular matrix degradation and vascular smooth muscle cell apoptosis are driven by cytokines. Previous studies emphasize an altered expression and a changed epigenetic regulation of key cytokines in AAA tissue samples. Such cytokines as IL-6, IL-10, IL-12, IL-17, IL-33, IL-1β, TGF-β, TNF-α, IFN-γ, and CXCL10 seem to be crucial in AAA pathogenesis. Some data obtained in animal studies show a protective function of IL-10, IL-33, and canonical TGF-β signaling, as well as a dual role of IL-4, IFN-γ and CXCL10, while TNF-α, IL-1β, IL-6, IL-12/IL-23, IL-17, CCR2, CXCR2, CXCR4 and the TGF-β noncanonical pathway are believed to aggravate the disease. Altogether data highlight significance of cytokines as informative markers and predictors of AAA. Pathologic serum/plasma concentrations of IL-1β, IL-2, IL-6, TNF-α, IL-10, IL-8, IL-17, IFN-γ, and PDGF have been already found in AAA patients. Some of the changes correlate with the size of aneurysms. Moreover, the risk of AAA is associated with polymorphic variants of genes encoding cytokines and their receptors: CCR2 (rs1799864), CCR5 (Delta-32), IL6 (rs1800796 and rs1800795), IL6R (rs12133641), IL10 (rs1800896), TGFB1 (rs1800469), TGFBR1 (rs1626340), TGFBR2 (rs1036095, rs4522809, rs1078985), and TNFA (rs1800629). Finally, 5 single-nucleotide polymorphisms in gene coding latent TGF-β-binding protein (LTBP4) and an allelic variant of TGFB3 are related to a significantly slower AAA annual growth rate.
Collapse
Affiliation(s)
- Olesya A Puchenkova
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Vladislav O Soldatov
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Andrei E Belykh
- Department of Pathophysiology, Research Institute of General Pathology, Kursk State Medical University, Kursk, Russia
- Dioscuri Centre for Metabolic Diseases, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - OlgaYu Bushueva
- Department of Biology, Medical Genetics and Ecology, Laboratory of Genomic Research at the Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
| | - Gennadii A Piavchenko
- Department of Histology, Cytology and Embryology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Laboratory of Cell Pathology in Critical State, State Research Institute of General Reanimatology, Moscow, Russia
| | - Artem A Venediktov
- Department of Histology, Cytology and Embryology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Alexey V Deykin
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Mikhail V Korokin
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Mikhail V Pokrovskiy
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| |
Collapse
|
7
|
Si K, Lu D, Tian J. Integrated analysis and the identification of a circRNA-miRNA-mRNA network in the progression of abdominal aortic aneurysm. PeerJ 2022; 9:e12682. [PMID: 35036156 PMCID: PMC8711282 DOI: 10.7717/peerj.12682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/02/2021] [Indexed: 11/20/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a disease commonly seen in the elderly. The aneurysm diameter increases yearly, and the larger the AAA the higher the risk of rupture, increasing the risk of death. However, there are no current effective interventions in the early stages of AAA. Methods Four gene expression profiling datasets, including 23 normal artery (NOR) tissue samples and 97 AAA tissue samples, were integrated in order to explore potential molecular biological targets for early intervention. After preprocessing, differentially expressed genes (DEGs) between AAA and NOR were identified using LIMMA package. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were conducted using the DAVID database. The protein-protein interaction network was constructed and hub genes were identified using the STRING database and plugins in Cytoscape. A circular RNA (circRNA) profile of four NOR tissues versus four AAA tissues was then reanalyzed. A circRNA-miRNA-mRNA interaction network was constructed after predictions were made using the Targetscan and Circinteractome databases. Results A total of 440 DEGs (263 up-regulated and 177 down-regulated) were identified in the AAA group, compared with the NOR group. The majority were associated with the extracellular matrix, tumor necrosis factor-α, and transforming growth factor-β. Ten hub gene-encoded proteins (namely IL6, RPS27A, JUN, UBC, UBA52, FOS, IL1B, MMP9, SPP1 and CCL2) coupled with a higher degree of connectivity hub were identified after protein‐protein interaction network analysis. Our results, in combination with the results of previous studies revealed that miR-635, miR-527, miR-520h, miR-938 and miR-518a-5p may be affected by circ_0005073 and impact the expression of hub genes such as CCL2, SPP1 and UBA52. The miR-1206 may also be affected by circ_0090069 and impact RPS27A expression. Conclusions This circRNA-miRNA-mRNA network may perform critical roles in AAA and may be a novel target for early intervention.
Collapse
Affiliation(s)
- Ke Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Da Lu
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai, People's Republic of China
| | - Jianbo Tian
- Institute of Information Engineering, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
8
|
Lv X, Hu Y, Chen X, Chen X, Chen L, Lin Y, Hou Y. Establishment and effect evaluation of an aortic dissection model induced by different doses of β-aminopropionitrile in rats. Vascular 2021; 29:832-840. [PMID: 33357159 DOI: 10.1177/1708538120984056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Given the controversy regarding the appropriate dose of β-aminopropionitrile for induction of aortic dissection models in rats, the purpose of this study was to explore the most suitable concentration of β-aminopropionitrile to establish a high-incidence and low-mortality aortic dissection model. METHODS Eighty three-week-old male Sprague-Dawley rats were equally divided into four groups: a control group, a 0.06% β-aminopropionitrile group, a 0.08% β-aminopropionitrile group and a 0.1% β-aminopropionitrile group. Initial experiments were performed on the control group, which was not treated with β-aminopropionitrile (and drank water freely), and the other three groups, which were given different concentrations of β-aminopropionitrile solution daily (0.06%, 0.08% and 0.1%). Subsequently, on the 40th day, osmotic minipumps administering 1 μg/kg per min angiotensin II (Ang II) were implanted subcutaneously into the β-aminopropionitrile groups, while the control group was continuously pumped with normal saline. The rats were euthanized 48 h after implantation. All rats that died before the expected end time of the experiment were autopsied immediately, and the aortas were dissected. The rats surviving at the end of the experiment were sacrificed by an overdose of sodium pentobarbital, and tissue samples were harvested for further analyses. RESULTS The mean survival days were significantly different among the groups, with 39.1 ± 6.04 days in the 0.08% β-aminopropionitrile group and 32.7 ± 9.85 days in the 0.1% β-aminopropionitrile group (P = 0.0178) at the end of the experiment. Compared with those in the 0.06% β-aminopropionitrile group, the rates of aortic dissection were significantly higher in the 0.08% β-aminopropionitrile group and the 0.1% β-aminopropionitrile group (P = 0.0015 and P = 0.0005, respectively), while there was no significant difference between the 0.08% β-aminopropionitrile group and the 0.1% β-aminopropionitrile group (P = 0.723) at 70% and 75%, respectively. However, the rupture rates were significantly different between the 0.08% β-aminopropionitrile group and the 0.1% β-aminopropionitrile group (55% versus 20%, P = 0.022). Hematoxylin-eosin staining of the aortic tissue sections of the β-aminopropionitrile group showed that red blood cells entered the pseudocavity in the vascular wall, while the vascular wall structure of the control group was intact. Compared with control rats, which were intact and free from fracture, β-aminopropionitrile-treated rats had fewer collagen fibers and exhibited fracture. Magnetic resonance imaging showed that the aortic intimae of the aortic dissection rats showed double lumens and intimal tears. CONCLUSIONS An aortic dissection model with a high incidence and low mortality was successfully and stably developed with 0.08% β-aminopropionitrile. This model will enable further studies investigating aortic dissection pathogenesis and drug therapy. Magnetic resonance imaging may be a reliable technique for imaging the aorta in rats.
Collapse
Affiliation(s)
- Xiaochai Lv
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yunnan Hu
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaodong Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xingfeng Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yong Lin
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yanting Hou
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
9
|
Abstract
Several studies have investigated the pathogenesis of aortic wall abnormalities such as aortic dissection or aneurysm; however, the comprehensive pathological in situ event involved in the development of the disease is not understood well. The vasa vasorum form a network of capillaries or venules around the adventitia and outer media, which play an important role in the aortic wall structure and function. Impairment of their function may induce tissue hypoxia, impede the transfer of cellular nutrients, and cause aortic medial degeneration, which is considered the major predisposing factor to this aortic wall pathology. This review updates our understanding of the pathological changes in the aortic media and vasa vasorum of patients with aortic dissection and aortic aneurysm.
Collapse
Affiliation(s)
- Hiroaki Osada
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
10
|
Chen SW, Chou SH, Tung YC, Hsiao FC, Ho CT, Chan YH, Wu VCC, Chou AH, Hsu ME, Lin PJ, Kao WWY, Chu PH. Expression and role of lumican in acute aortic dissection: A human and mouse study. PLoS One 2021; 16:e0255238. [PMID: 34310653 PMCID: PMC8312931 DOI: 10.1371/journal.pone.0255238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
Introduction Aortic dissection (AD) is a life-threatening emergency, and lumican (LUM) is a potential Biomarker for AD diagnosis. We investigated LUM expression patterns in patients with AD and explored the molecular functions of Lum in AD mice model. Methods LUM expression patterns were analyzed using aortic tissues of AD patients, and serum soluble LUM (s-LUM) levels were compared between patients with acute AD (AAD) and chronic AD (CAD). Lum-knockout (Lum−/−) mice were challenged with β-aminopropionitrile (BAPN) and angiotensin II (Ang II) to induce AD. The survival rate, AD incidence, and aortic aneurysm (AA) in these mice were compared with those in BAPN–Ang II–challenged wildtype (WT) mice. Tgf-β/Smad2, Mmps, Lum, and Nox expression patterns were examined. Results LUM expression was detected in the intima and media of the ascending aorta in patients with AAD. Serum s-LUM levels were significantly higher in patients with AAD than CAD. Furthermore, AD-associated mortality and thoracic aortic rupture incidence were significantly higher in the Lum−/− AD mice than in the WT AD mice. However, no significant pathologic changes in AA were observed in the Lum−/− AD mice compared with the WT AD mice. The BAPN–Ang II–challenged WT and Lum−/− AD mice had higher Tgf-β, p-Smad2, Mmp2, Mmp9, and Nox4 levels than those of non-AD mice. We also found that Lum expression was significantly higher in the BAPN-Ang II–challenged WT in comparison to the unchallenged WT mice. Conclusion LUM expression was altered in patients with AD display increased s-LUM in blood, and Lum−/− mice exhibited augmented AD pathogenesis. These findings support the notion that LUM is a biomarker signifying the pathogenesis of injured aorta seen in AAD. The presence of LUM is essential for maintenance of connective tissue integrity. Future studies should elucidate the mechanisms underlying LUM association in aortic changes.
Collapse
Affiliation(s)
- Shao-Wei Chen
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City, Taiwan
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Shing-Hsien Chou
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Ying-Chang Tung
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Fu-Chih Hsiao
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Chien-Te Ho
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Yi-Hsin Chan
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - Victor Chien-Chia Wu
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
| | - An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City, Taiwan
| | - Ming-En Hsu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City, Taiwan
| | - Pyng-Jing Lin
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan City, Taiwan
| | - Winston W. Y. Kao
- Crawley Vision Research Center, Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Pao-Hsien Chu
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- * E-mail:
| |
Collapse
|
11
|
Zhao B, Sun Q, Fan Y, Hu X, Li L, Wang J, Cui S. Transplantation of bone marrow-derived mesenchymal stem cells with silencing of microRNA-138 relieves pelvic organ prolapse through the FBLN5/IL-1β/elastin pathway. Aging (Albany NY) 2021; 13:3045-3059. [PMID: 33460398 PMCID: PMC7880387 DOI: 10.18632/aging.202465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
Nondegradable transvaginal polypropylene meshes for treating pelvic organ prolapse (POP) are now generally unavailable or banned due to serious adverse events. New tissue engineering approaches combine degradable scaffolds with mesenchymal stem/stromal cells from human endometrium (eMSC). In this study, we investigate effect of microRNA-138 (miR-138) regulation on bone marrow-derived mesenchymal stem cells (BMSCs) and the efficacy of BMSC transplantation therapy in a rat POP model. We first identified FBLN5 as a target of miR-138. miR-138, fibulin-5 (FBLN5), interleukin-1β (IL-1β), and elastin expression in uterosacral ligament of POP patients and controls were detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. After isolation and identification, BMSCs were treated to alter their expression of miR-138 or FBLN5. Proliferation of BMSCs was analyzed by CCK-8. After establishing the rat pelvic floor dysfunction (PFD) model, we evaluated efficacy of BMSC injection by applying leak point pressure (LPP) and the conscious cystometry (CMG) tests. miR-138 inhibition resulted in increased viability of BMSCs and elevated their secretion of elastin, while downregulating IL-1β expression. BMSCs with inhibited miR-138 improved LPP and conscious CMG results in vivo. Taken together, miR-138 could be a potential therapeutic target for treating POP in conjunction with tissue engineering.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Qing Sun
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Yazhou Fan
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Xinming Hu
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Linyu Li
- Department of Scientific Research, Xinxiang Medical University Sanquan Medical College, Xinxiang 453003, Henan Province, PR China
| | - Junmin Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, PR China
| | - Shihong Cui
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| |
Collapse
|
12
|
Re "Deciphering the Role of Interleukin-1β in the Development of Dissecting Thoracic Aortic Aneurysm". Eur J Vasc Endovasc Surg 2020; 61:348-349. [PMID: 33262089 DOI: 10.1016/j.ejvs.2020.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 11/23/2022]
|
13
|
Lareyre F, Raffort J. Deciphering the Role of Interleukin-1β in the Development of Dissecting Thoracic Aortic Aneurysm. Eur J Vasc Endovasc Surg 2020; 61:348. [PMID: 33281025 DOI: 10.1016/j.ejvs.2020.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 11/15/2022]
Affiliation(s)
- Fabien Lareyre
- Department of Vascular Surgery, Hospital of Antibes Juan-les-Pins, Antibes, France; Université Côte d'Azur, CHU, Inserm U1065, C3M, Nice, France.
| | - Juliette Raffort
- Université Côte d'Azur, CHU, Inserm U1065, C3M, Nice, France; Clinical Chemistry Laboratory, University Hospital of Nice, France
| |
Collapse
|
14
|
Malecki C, Hambly BD, Jeremy RW, Robertson EN. The Role of Inflammation and Myeloperoxidase-Related Oxidative Stress in the Pathogenesis of Genetically Triggered Thoracic Aortic Aneurysms. Int J Mol Sci 2020; 21:ijms21207678. [PMID: 33081376 PMCID: PMC7590002 DOI: 10.3390/ijms21207678] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Genetically triggered thoracic aortic aneurysms (TAAs) are usually considered to exhibit minimal levels of inflammation. However, emerging data demonstrate that specific features of an inflammatory response can be observed in TAA, and that the extent of the inflammatory response can be correlated with the severity, in both mouse models and in human studies. Myeloperoxidase (MPO) is a key mediator of the inflammatory response, via production of specific oxidative species, e.g., the hypohalous acids. Specific tissue modifications, mediated by hypohalous acids, have been documented in multiple cardiovascular pathologies, including atherosclerosis associated with coronary artery disease, abdominal aortic, and cerebral aneurysms. Similarly, data are now emerging that show the capacity of MPO-derived oxidative species to regulate mechanisms important in TAA pathogenesis, including alterations in extracellular matrix homeostasis, activation of matrix metalloproteinases, induction of endothelial dysfunction and vascular smooth muscle cell phenotypic switching, and activation of ERK1/2 signaling. The weight of evidence supports a role for inflammation in exacerbating the severity of TAA progression, expanding our understanding of the pathogenesis of TAA, identifying potential biomarkers for early detection of TAA, monitoring severity and progression, and for defining potential novel therapeutic targets.
Collapse
Affiliation(s)
- Cassandra Malecki
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
- Correspondence:
| | - Brett D. Hambly
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
| | - Richmond W. Jeremy
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Elizabeth N. Robertson
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| |
Collapse
|
15
|
Guo LL, Wu MT, Zhang LW, Chu YX, Tian P, Jing ZP, Li JS, Sun YD, Yeung KK, Zhang L. Blocking Interleukin-1 Beta Reduces the Evolution of Thoracic Aortic Dissection in a Rodent Model. Eur J Vasc Endovasc Surg 2020; 60:916-924. [PMID: 33004280 DOI: 10.1016/j.ejvs.2020.08.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 07/28/2020] [Accepted: 08/20/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Thoracic aortic dissection (TAD) is associated with matrix changes, biochemical changes, and inflammatory markers like interleukin-1 beta (IL-1β). However, the exact mechanism remains unknown. This study aimed to investigate the role of IL-1β, matrix metalloproteinase (MMP)-2, MMP-9, smooth muscle cell apoptosis, and elastic fibre fracture in the development of TAD in a rat model. METHODS The TAD rat model was induced by β-aminopropionitrile (BAPN). TAD was investigated in 112 male Sprague-Dawley rats, which were equally divided into four groups of 28 rats (Control, BAPN, BAPN + IL-1β, and BAPN + IL-1β antibody). Systolic blood pressure, survival, and the development of TAD were measured after six weeks. Expression of IL-1β, MMP-2, and MMP-9 was measured by Western blot. Apoptosis, aortic elastin concentration, and biomechanical characteristics were measured by the TdT mediated dUTP nick end labelling assay, Victoria blue staining, and in vitro testing. RESULTS During six weeks, the mortality was 0% (0/28) in the control group, 53.6% (15/28) in the BAPN group (p < .001 compared with the control group), 75.0% (21/28) in the BAPN + IL-1β group (p = .007 compared with the BAPN group), and 35.7% (10/28) in the BAPN + IL-1β antibody group (p = .023 compared with BAPN group and p < .001 compared with the BAPN + IL-1β group). IL-1β treatment deteriorates BAPN induced mortality and aneurysm expansion, which were attenuated by anti-IL-1β treatment. In BAPN + IL-1β group, stress and strain parameters were decreased by 13.5%-53.5% and elastin content was decreased by 14%, and IL-1β, MMP-2, and MMP-9 were expressed higher by 117%, 108%, and 75% when compared with the rats in the BAPN group. Contrarily, in the BAPN + IL-1β antibody group, the above changes could be completely (strain, elastin content, and expression of MMP-2) or partly (elasticity modulus, stress, and expression of MMP-9) blocked by anti-IL-1β treatment. CONCLUSION IL-1β plays a critical role in TAD formation by altering the expression of MMP-2 and MMP-9, degrading the aortic wall matrix, causing elastic fibre rupture, and changing the stress or strain of the aortic wall. Anti-IL-1β reduces the later effects and could be one of the molecular targets for prognosis and drug treatment of TAD in the future.
Collapse
Affiliation(s)
- Ling-Ling Guo
- Department of Biological Therapies for Cancer, Eastern Hepatobiliary Surgery Hospital, Navy (Second) Military Medical University, Shanghai, China
| | - Meng-Tao Wu
- Department of Vascular Surgery, The Second Hospital of Shandong University, Ji'nan, PR China
| | - Li-Wei Zhang
- Department of Cardio-Thoracic Surgery, The People's Hospital of China Three Gorges University, Yichang, China
| | - Yong-Xin Chu
- Department of Vascular Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Peng Tian
- Department of Vascular Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China.
| | - Zai-Ping Jing
- Department of Vascular Surgery, Changhai Hospital, Navy (Second) Military Medical University, Shanghai, China
| | - Jia-Si Li
- Department of Neurology, Changhai Hospital, Navy (Second) Military Medical University, Shanghai, China
| | - Yu-Dong Sun
- Department of Vascular Surgery, Changhai Hospital, Navy (Second) Military Medical University, Shanghai, China
| | - Kak K Yeung
- Department of Vascular Surgery, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Lei Zhang
- Department of Vascular Surgery, Changhai Hospital, Navy (Second) Military Medical University, Shanghai, China.
| |
Collapse
|
16
|
Abstract
Mutations in extracellular matrix and smooth muscle cell contractile proteins predispose to thoracic aortic aneurysms in Marfan syndrome (MFS) and related disorders. These genetic alterations lead to a compromised extracellular matrix-smooth muscle cell contractile unit. The abnormal aortic tissue responds with defective mechanosensing under hemodynamic stress. Aberrant mechanosensing is associated with transforming growth factor-beta (TGF-β) hyperactivity, enhanced angiotensin-II (Ang-II) signaling, and perturbation of other cellular signaling pathways. The downstream consequences include enhanced proteolytic activity, expression of inflammatory cytokines and chemokines, infiltration of inflammatory cells in the aortic wall, vascular smooth muscle cell apoptosis, and medial degeneration. Mouse models highlight aortic inflammation as a contributing factor in the development of aortic aneurysms. Anti-inflammatory drugs and antioxidants can reduce aortic oxidative stress that prevents aggravation of aortic disease in MFS mice. Targeting TGF-β and Ang-II downstream signaling pathways such as ERK1/2, mTOR, PI3/Akt, P38/MAPK, and Rho kinase signaling attenuates disease pathogenesis. Aortic extracellular matrix degradation and medial degeneration were reduced upon inhibition of inflammatory cytokines and matrix metalloproteinases, but the latter lack specificity. Treating inflammation associated with aortic aneurysms in MFS and related disorders could prove to be beneficial in limiting disease pathogenesis.
Collapse
|